1
|
Navarro-Bailón A, López-Parra M, Veiga-Vaz Á, Villarón EM, Díez-Campelo M, Martín AÁ, Pérez-López E, Cabrero M, Vázquez L, López-Corral L, Sánchez-Guijo F. Treatment of post-allogeneic hematopoietic stem cell transplant cytopenias with sequential doses of multipotent mesenchymal stromal/stem cells. Cytotherapy 2024; 26:806-812. [PMID: 38727653 DOI: 10.1016/j.jcyt.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND AIMS Cytopenias after allogeneic stem cell transplantation (allo-SCT) are a common complication, the underlying pathogenic mechanisms of which remain incompletely understood. Multipotent mesenchymal stromal/stem cell (MSC) therapy has been successfully employed in the treatment of immune-related disorders and can aid in the restoration of the hematopoietic niche. METHODS A phase II clinical trial to assess the efficacy and safety of administering four sequential doses of ex-vivo expanded bone marrow MSCs from a third-party donor to patients with persistent severe cytopenias after allo-SCT was performed. RESULTS The overall response rate on day 90 was 75% among the 27 evaluable patients (comprising 12 complete responses, 8 partial responses, and 7 with no response). The median time to respond was 14.5 days. Responses were observed across different profiles, including single or multiple affected lineages, primary or secondary timing, and potential immune-mediated or post-infectious pathophysiology versus idiopathic origin. With a median follow-up for surviving patients of 85 months after MSC infusion, 53% of patients are alive. Notably, no adverse events related to MSC therapy were reported. CONCLUSIONS In summary, the sequential infusion of third-party MSCs emerges as a viable and safe therapeutic option, exhibiting potential benefits for patients experiencing cytopenias following allo-SCT.
Collapse
Affiliation(s)
- Almudena Navarro-Bailón
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain.
| | - Miriam López-Parra
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain
| | - Álvaro Veiga-Vaz
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Eva María Villarón
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain
| | - María Díez-Campelo
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain; Biomedical Research Networking Center for Cancer (CIBERONC), ISCIII, Spain
| | - Ana África Martín
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Estefanía Pérez-López
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Mónica Cabrero
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Lourdes Vázquez
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Lucía López-Corral
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain; Biomedical Research Networking Center for Cancer (CIBERONC), ISCIII, Spain
| | - Fermín Sánchez-Guijo
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain; Biomedical Research Networking Center for Cancer (CIBERONC), ISCIII, Spain
| |
Collapse
|
2
|
Wu SCM, Zhu M, Chik SCC, Kwok M, Javed A, Law L, Chan S, Boheler KR, Liu YP, Chan GCF, Poon ENY. Adipose tissue-derived human mesenchymal stromal cells can better suppress complement lysis, engraft and inhibit acute graft-versus-host disease in mice. Stem Cell Res Ther 2023; 14:167. [PMID: 37357314 DOI: 10.1186/s13287-023-03380-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 05/18/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Acute graft-versus-host disease (aGvHD) is a life-threatening complication of allogeneic hematopoietic stem cell transplantation (HSCT). Transplantation of immunosuppressive human mesenchymal stromal cells (hMSCs) can protect against aGvHD post-HSCT; however, their efficacy is limited by poor engraftment and survival. Moreover, infused MSCs can be damaged by activated complement, yet strategies to minimise complement injury of hMSCs and improve their survival are limited. METHODS Human MSCs were derived from bone marrow (BM), adipose tissue (AT) and umbilical cord (UC). In vitro immunomodulatory potential was determined by co-culture experiments between hMSCs and immune cells implicated in aGvHD disease progression. BM-, AT- and UC-hMSCs were tested for their abilities to protect aGvHD in a mouse model of this disease. Survival and clinical symptoms were monitored, and target tissues of aGvHD were examined by histopathology and qPCR. Transplanted cell survival was evaluated by cell tracing and by qPCR. The transcriptome of BM-, AT- and UC-hMSCs was profiled by RNA-sequencing. Focused experiments were performed to compare the expression of complement inhibitors and the abilities of hMSCs to resist complement lysis. RESULTS Human MSCs derived from three tissues divergently protected against aGvHD in vivo. AT-hMSCs preferentially suppressed complement in vitro and in vivo, resisted complement lysis and survived better after transplantation when compared to BM- and UC-hMSCs. AT-hMSCs also prolonged survival and improved the symptoms and pathological features of aGvHD. We found that complement-decay accelerating factor (CD55), an inhibitor of complement, is elevated in AT-hMSCs and contributed to reduced complement activation. We further report that atorvastatin and erlotinib could upregulate CD55 and suppress complement in all three types of hMSCs. CONCLUSION CD55, by suppressing complement, contributes to the improved protection of AT-hMSCs against aGvHD. The use of AT-hMSCs or the upregulation of CD55 by small molecules thus represents promising new strategies to promote hMSC survival to improve the efficacy of transplantation therapy. As complement injury is a barrier to all types of hMSC therapy, our findings are of broad significance to enhance the use of hMSCs for the treatment of a wide range of disorders.
Collapse
Affiliation(s)
- Stanley Chun Ming Wu
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Manyu Zhu
- Department of Orthopaedics and Traumatology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Stanley C C Chik
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Maxwell Kwok
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong, Kowloon Bay, Hong Kong SAR, China
| | - Asif Javed
- School of Biomedical Science, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Laalaa Law
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Shing Chan
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kenneth R Boheler
- Division of Cardiology, Department of Medicine and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yin Ping Liu
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Godfrey Chi Fung Chan
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
- , Doctors' Office, 9/F, Tower B, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Hong Kong SAR, China.
| | - Ellen Ngar-Yun Poon
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong, Kowloon Bay, Hong Kong SAR, China.
- The School of Biomedical Sciences, The Chinese University of Hong Kong, Rm 226A, 2/F, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Shatin, Hong Kong SAR, China.
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
3
|
Hong T, Wang R, Yang G, Wang X, Zeng L, Yang S, Wei J, Gao Q, Zhang X. Human umbilical cord mesenchymal stem cells ameliorate acute graft versus host disease by elevating phytosphingosine. Exp Hematol 2023:S0301-472X(23)00070-X. [PMID: 36931619 DOI: 10.1016/j.exphem.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a prominent barrier to allogeneic hematopoietic stem cell transplantation (allo-HSCT) and even leads to death after HSCT. Human umbilical cord mesenchymal stem cells (HUCMSCs) are effective in aGVHD treatment and have mild side effects, but the underlying mechanisms remain unclear. Phytosphingosine (PHS) is known to prevent loss of moisture from the skin; regulate epidermal cell growth, differentiation, and apoptosis; and exert bactericidal and anti-inflammatory effects. In this study, our results revealed the efficacy of HUCMSCs in alleviating aGVHD in a murine model, with striking changes in metabolism and significantly elevated PHS levels due to sphingolipid metabolism. In vitro, PHS reduced CD4+ T cell proliferation, enhanced apoptosis and reduced T helper 1 (Th1) cell differentiation. Transcriptional analysis of donor CD4+ T cells treated with PHS revealed significant decreases in transcripts regulating proinflammatory pathways, such as NF-κB. In vivo, the administration of PHS significantly ameliorated aGVHD development. Collectively, these beneficial effects indicate proof-of-concept that sphingolipid metabolites could be a safe and effective means to prevent aGVHD in the clinic.
Collapse
Affiliation(s)
- Tao Hong
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Rui Wang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China
| | - Guancui Yang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Lingyu Zeng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221008, China
| | - Shijie Yang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China
| | - Jin Wei
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Qiangguo Gao
- Department of Cell Biology, College of Basic Medicine, Army Medical University, Chongqing, 400038, China..
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital. State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China..
| |
Collapse
|
4
|
Kidpun P, Ruanglertboon W, Chalongsuk R. State-of-the-art knowledge on the regulation of advanced therapy medicinal products. Per Med 2022; 19:251-261. [PMID: 35293224 DOI: 10.2217/pme-2021-0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Advanced therapy medicinal products (ATMPs) constitute therapeutic agents based on obtained cells, tissues or genes representing a novel treatment opportunity in medicine. In addition, ATMPs are administered into the cells or tissues of humans from the patient's own cells, donors, or genetically modified cells. Recently, the field of developing ATMPs has become a point of attention due to the clinical efficacy expected in defeating incurable diseases such as cancers and neurodegenerative disorders. Currently, there are two modes regarding the distribution of ATMPs. First, ATMPs that might be legally authorized for marketing. Second, the patients are able to access unapproved ATMPs through the hospital exemption (HE) or clinical practice program or through the compassionate use and expanded access program. The aim of this review is to discuss state-of-the-art knowledge on the regulation of ATMPs and provide regulatory recommendations.
Collapse
Affiliation(s)
- Patcharaphun Kidpun
- Department of Community Pharmacy, Faculty of Pharmacy, Silpakorn University, Sanam Chandra Palace Campus, Nakhon Pathom, Thailand
| | - Warit Ruanglertboon
- Discipline of Pharmacology, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Rapeepun Chalongsuk
- Department of Community Pharmacy, Faculty of Pharmacy, Silpakorn University, Sanam Chandra Palace Campus, Nakhon Pathom, Thailand
| |
Collapse
|
5
|
A phase 2a randomized clinical trial of intravenous vedolizumab for the treatment of steroid-refractory intestinal acute graft-versus-host disease. Bone Marrow Transplant 2021; 56:2477-2488. [PMID: 34108672 PMCID: PMC8486663 DOI: 10.1038/s41409-021-01356-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 04/23/2021] [Accepted: 05/12/2021] [Indexed: 01/10/2023]
Abstract
Steroid-refractory (SR) acute graft-versus-host disease (aGvHD) remains a significant complication after allogeneic hematopoietic cell transplantation. Systemic corticosteroids are first-line therapy for aGvHD, but apart from ruxolitinib, there are no approved treatments for SR aGvHD. Vedolizumab is approved for treatment of ulcerative colitis and Crohn’s disease, and may be effective for treatment of SR intestinal aGvHD. We conducted a phase 2a trial (NCT02993783) to evaluate the clinical efficacy, tolerability, and safety of vedolizumab 300 and 600 mg for SR intestinal aGvHD. This study was terminated before full enrollment was completed because early results failed to demonstrate positive proof-of-concept in efficacy. Before termination, 17 participants had enrolled and an early response in intestinal aGvHD was observed in 11 and eight participants at days 15 and 28, respectively. All adverse events observed were consistent with those expected in a population with SR intestinal aGvHD. Overall, vedolizumab did not meet the primary efficacy endpoint (overall response at day 28), likely owing to premature study drug discontinuation, lack of efficacy, and the competing risks inherent with a population with advanced SR intestinal aGvHD. Nevertheless, this study provides valuable insights into the considerations needed when conducting studies in patients with SR intestinal aGvHD.
Collapse
|
6
|
Lechanteur C, Briquet A, Bettonville V, Baudoux E, Beguin Y. MSC Manufacturing for Academic Clinical Trials: From a Clinical-Grade to a Full GMP-Compliant Process. Cells 2021; 10:1320. [PMID: 34073206 PMCID: PMC8227789 DOI: 10.3390/cells10061320] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Following European regulation 1394/2007, mesenchymal stromal cell (MSCs) have become an advanced therapy medicinal product (ATMP) that must be produced following the good manufacturing practice (GMP) standards. We describe the upgrade of our existing clinical-grade MSC manufacturing process to obtain GMP certification. Staff organization, premises/equipment qualification and monitoring, raw materials management, starting materials, technical manufacturing processes, quality controls, and the release, thawing and infusion were substantially reorganized. Numerous studies have been carried out to validate cultures and demonstrate the short-term stability of fresh or thawed products, as well their stability during long-term storage. Detailed results of media simulation tests, validation runs and early MSC batches are presented. We also report the validation of a new variant of the process aiming to prepare fresh MSCs for the treatment of specific lesions of Crohn's disease by local injection. In conclusion, we have successfully ensured the adaptation of our clinical-grade MSC production process to the GMP requirements. The GMP manufacturing of MSC products is feasible in the academic setting for a limited number of batches with a significant cost increase, but moving to large-scale production necessary for phase III trials would require the involvement of industrial partners.
Collapse
Affiliation(s)
- Chantal Lechanteur
- Laboratory of Cell and Gene Therapy, Department of Hematology, CHU of Liège, 4000 Liège, Belgium; (A.B.); (V.B.); (E.B.); (Y.B.)
| | - Alexandra Briquet
- Laboratory of Cell and Gene Therapy, Department of Hematology, CHU of Liège, 4000 Liège, Belgium; (A.B.); (V.B.); (E.B.); (Y.B.)
| | - Virginie Bettonville
- Laboratory of Cell and Gene Therapy, Department of Hematology, CHU of Liège, 4000 Liège, Belgium; (A.B.); (V.B.); (E.B.); (Y.B.)
| | - Etienne Baudoux
- Laboratory of Cell and Gene Therapy, Department of Hematology, CHU of Liège, 4000 Liège, Belgium; (A.B.); (V.B.); (E.B.); (Y.B.)
| | - Yves Beguin
- Laboratory of Cell and Gene Therapy, Department of Hematology, CHU of Liège, 4000 Liège, Belgium; (A.B.); (V.B.); (E.B.); (Y.B.)
- Division of Hematology, Department of Medicine, CHU of Liège, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
7
|
Hansen M, Stahl L, Heider A, Hilger N, Sack U, Kirschner A, Cross M, Fricke S. Reduction of Graft-versus-Host-Disease in NOD.Cg-Prkdc scid Il2rg tm1Wjl/SzJ (NSG) Mice by Cotransplantation of Syngeneic Human Umbilical Cord-Derived Mesenchymal Stromal Cells. Transplant Cell Ther 2021; 27:658.e1-658.e10. [PMID: 33964513 DOI: 10.1016/j.jtct.2021.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/16/2021] [Accepted: 04/25/2021] [Indexed: 12/12/2022]
Abstract
Graft-versus-host disease (GVHD) is one of the major complications following hematopoietic stem cell transplantation, which remains the sole curative therapy for many malignant diseases of the hematopoietic system. The immunomodulatory potential of mesenchymal stromal cells (MSCs) to treat GVHD is currently being tested in various preclinical and clinical trials. Because the results of the preclinical and clinical trials on the use of MSCs to treat GVHD have not been consistent, we analyzed the potential beneficial effects of syngeneic versus allogenic treatment, culture expansion of MSCs, and various MSC cell doses and time points of MSC transplantation in a murine GVHD model. We established the murine GVHD model based on the transplantation of umbilical cord blood-derived hematopoietic stem cells (UC-HSCs) and used this model to assess the therapeutic potential of umbilical cord blood-derived MSCs (UC-MSCs). The use of HSC and MSC populations derived from the same donor allowed us to exclude third-party cells and test the UC-HSCs and UC-MSCs in a matched setting. Moreover, we were able to compare various doses, transplantation time points, and the influence of culture expansion of MSCs on the impact of treatment. This resulted in 16 different treatment groups. The most efficient setting for treatment of UC-HSC-induced GVHD reactions was based on the simultaneous administration of 1 × 106 culture-expanded, syngeneically matched UC-MSCs. This therapy effectively reduced the number of CD8+ T cells in the blood, protected the mice from weight loss, and prolonged their survival until the end of observation period. Taken together, our data show beneficial effects of (1) syngeneic over allogeneic UC-HSCs and UC-MSCs, (2) culture-expanded cells over freshly isolated primary cells, (3) simultaneous over sequential administration, and (4) high doses of UC-MSCs. The animal model of GVHD established here is now available for more detailed studies, including a comparative analysis of the efficacy of MSCs derived from alternative sources, such as adipose tissue and bone marrow.
Collapse
Affiliation(s)
- Max Hansen
- Vita 34 AG, Leipzig, Germany; Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany.
| | - Lilly Stahl
- Tcell Tolerance GmbH, Leipzig, Germany; Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | | | - Nadja Hilger
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ulrich Sack
- Department of Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Andreas Kirschner
- Vita 34 AG, Leipzig, Germany; Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Michael Cross
- Department of Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
8
|
Gruhn B, Brodt G, Ernst J. Extended Treatment With Mesenchymal Stromal Cells-Frankfurt am Main in a Pediatric Patient With Steroid-refractory Acute Gastrointestinal Graft-Versus-Host Disease: Case Report and Review of the Literature. J Pediatr Hematol Oncol 2021; 43:e419-e425. [PMID: 32118816 DOI: 10.1097/mph.0000000000001758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/30/2020] [Indexed: 12/14/2022]
Abstract
In acute graft-versus-host disease (aGVHD) following allogeneic hematopoietic stem cell transplantation, there are various options available after the failure of initial steroid therapy. Since the publication of the first study in 2008, mesenchymal stromal cells (MSCs) have also been used with increasing frequency, including in pediatric patients with steroid-refractory aGVHD, and the manufacturing process has undergone further development. MSC-Frankfurt am Main (MSC-FFM, Obnitix), which is manufactured from pooled mononuclear bone marrow cells from 8 donors using a standardized process, resulted in a response rate of 84% in children with steroid-refractory aGVHD. We report on a 13-year-old female patient with acute myeloid leukemia who received Obnitix as a third-line treatment for gastrointestinal (GI) aGVHD in a life-threatening situation. The patient was initially given a total of 4 Obnitix infusions as per the regulatory approval, with her symptoms improving from day 9 after the first infusion. The second cycle of 4 Obnitix infusions followed due to persistent severe protein-losing enteropathy and resulted in complete remission. A systematic review of the literature on MSC in pediatric patients with steroid-refractory aGVHD confirms that MSC treatment beyond 4 weeks is employed in accordance with treatment protocols or on a case-by-case basis. To summarize, aGVHD activity can be checked endoscopically in patients with persistent GI symptoms and a second Obnitix cycle can then be administered if appropriate, with the goal of achieving complete remission. Future studies should also investigate the potential influence of tissue repair properties as an element in MSCs' efficacy in GI aGVHD.
Collapse
Affiliation(s)
- Bernd Gruhn
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | | | | |
Collapse
|
9
|
Hamdan H, Hashmi SK, Lazarus H, Gale RP, Qu W, El Fakih R. Promising role for mesenchymal stromal cells in coronavirus infectious disease-19 (COVID-19)-related severe acute respiratory syndrome? Blood Rev 2021; 46:100742. [PMID: 32854985 PMCID: PMC7425550 DOI: 10.1016/j.blre.2020.100742] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/02/2020] [Accepted: 08/06/2020] [Indexed: 12/25/2022]
Abstract
Mesenchymal stromal cells (MSC) have immune regulatory and tissue regenerative properties. MSCs are being studied as a therapy option for many inflammatory and immune disorders and are approved to treat acute graft-versus-host disease (GvHD). The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic and associated coronavirus infectious disease-19 (COVID-19) has claimed many lives. Innovative therapies are needed. Preliminary data using MSCs in the setting of acute respiratory distress syndrome (ARDS) in COVID-19 are emerging. We review mechanisms of action of MSCs in inflammatory and immune conditions and discuss a potential role in persons with COVID-19.
Collapse
Affiliation(s)
- Hamdan Hamdan
- Department of Physiology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia,Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shahrukh K. Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA,Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hillard Lazarus
- Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Wenchun Qu
- Department of Pain Medicine, Mayo Clinic, Jacksonville, FL, USA,Center of Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Riad El Fakih
- Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia,Corresponding author at: Oncology Centre, KFSHRC, Section of Adult Hematology/HSCT, PO Box 3354, Riyadh 11471, Saudi Arabia
| |
Collapse
|
10
|
Johnstone BH, Messner F, Brandacher G, Woods EJ. A Large-Scale Bank of Organ Donor Bone Marrow and Matched Mesenchymal Stem Cells for Promoting Immunomodulation and Transplant Tolerance. Front Immunol 2021; 12:622604. [PMID: 33732244 PMCID: PMC7959805 DOI: 10.3389/fimmu.2021.622604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Induction of immune tolerance for solid organ and vascular composite allografts is the Holy Grail for transplantation medicine. This would obviate the need for life-long immunosuppression which is associated with serious adverse outcomes, such as infections, cancers, and renal failure. Currently the most promising means of tolerance induction is through establishing a mixed chimeric state by transplantation of donor hematopoietic stem cells; however, with the exception of living donor renal transplantation, the mixed chimerism approach has not achieved durable immune tolerance on a large scale in preclinical or clinical trials with other solid organs or vascular composite allotransplants (VCA). Ossium Health has established a bank of cryopreserved bone marrow (BM), termed "hematopoietic progenitor cell (HPC), Marrow," recovered from deceased organ donor vertebral bodies. This new source for hematopoietic cell transplant will be a valuable resource for treating hematological malignancies as well as for inducing transplant tolerance. In addition, we have discovered and developed a large source of mesenchymal stem (stromal) cells (MSC) tightly associated with the vertebral body bone fragment byproduct of the HPC, Marrow recovery process. Thus, these vertebral bone adherent MSC (vBA-MSC) are matched to the banked BM obtained from each donor, as opposed to third-party MSC, which enhances safety and potentially efficacy. Isolation and characterization of vBA-MSC from over 30 donors has demonstrated that the cells are no different than traditional BM-MSC; however, their abundance is >1,000-fold higher than obtainable from living donor BM aspirates. Based on our own unpublished data as well as reports published by others, MSC facilitate chimerism, especially at limiting hematopoietic stem and progenitor cell (HSPC) numbers and increase safety by controlling and/or preventing graft-vs.-host-disease (GvHD). Thus, vBA-MSC have the potential to facilitate mixed chimerism, promote complementary peripheral immunomodulatory functions and increase safety of BM infusions. Both HPC, Marrow and vBA-MSC have potential use in current VCA and solid organ transplant (SOT) tolerance clinical protocols that are amenable to "delayed tolerance." Current trials with HPC, Marrow are planned with subsequent phases to include vBA-MSC for tolerance of both VCA and SOT.
Collapse
Affiliation(s)
- Brian H. Johnstone
- Ossium Health, Indianapolis, IN, United States
- Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, United States
| | - Franka Messner
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Erik J. Woods
- Ossium Health, Indianapolis, IN, United States
- Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
11
|
Cellular therapies for graft-versus-host disease: a tale of tissue repair and tolerance. Blood 2021; 136:410-417. [PMID: 32525970 DOI: 10.1182/blood.2019000951] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
The success of allogeneic hematopoietic cell transplantation depends heavily on the delicate balance between the activity of the donor immune system against malignant and nonmalignant cells of the recipient. Abrogation of alloreactivity will lead to disease relapse, whereas untamed allo-immune responses will lead to lethal graft-versus-host disease (GVHD). A number of cell types have been identified that can be used to suppress alloreactive immune cells and prevent lethal GVHD in mice. Of those, mesenchymal stromal cells and, to a lesser extent, regulatory T cells have demonstrated efficacy in humans. Ideally, cellular therapy for GVHD will not affect alloreactive immune responses against tumor cells. The importance of tissue damage in the pathophysiology of GVHD rationalizes the development of cells that support tissue homeostasis and repair, such as innate lymphoid cells. We discuss recent developments in the field of cellular therapy to prevent and treat acute and chronic GVHD, in the context of GVHD pathophysiology.
Collapse
|
12
|
The Potential of Mesenchymal Stromal Cells in Neuroblastoma Therapy for Delivery of Anti-Cancer Agents and Hematopoietic Recovery. J Pers Med 2021; 11:jpm11030161. [PMID: 33668854 PMCID: PMC7996318 DOI: 10.3390/jpm11030161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is one of the most common pediatric cancers and a major cause of cancer-related death in infancy. Conventional therapies including high-dose chemotherapy, stem cell transplantation, and immunotherapy approach a limit in the treatment of high-risk neuroblastoma and prevention of relapse. In the last two decades, research unraveled a potential use of mesenchymal stromal cells in tumor therapy, as tumor-selective delivery vehicles for therapeutic compounds and oncolytic viruses and by means of supporting hematopoietic stem cell transplantation. Based on pre-clinical and clinical advances in neuroblastoma and other malignancies, we assess both the strong potential and the associated risks of using mesenchymal stromal cells in the therapy for neuroblastoma. Furthermore, we examine feasibility and safety aspects and discuss future directions for harnessing the advantageous properties of mesenchymal stromal cells for the advancement of therapy success.
Collapse
|
13
|
Döring M, Cabanillas Stanchi KM, Lenglinger K, Treuner C, Gieseke F, Erbacher A, Mezger M, Vaegler M, Schlegel PG, Greil J, Bettoni da Cunha Riehm C, Faul C, Schumm M, Lang P, Handgretinger R, Müller I. Long-Term Follow-Up After the Application of Mesenchymal Stromal Cells in Children and Adolescents with Steroid-Refractory Graft-Versus-Host Disease. Stem Cells Dev 2021; 30:234-246. [PMID: 33446053 DOI: 10.1089/scd.2020.0191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Steroid-refractory graft-versus-host disease (GvHD) is a life-threatening complication after allogeneic hematopoietic stem cell transplantation (alloHSCT). Alternative treatment options are often insufficient. Several studies have proven the efficacy of mesenchymal stromal cells (MSCs) in the treatment of therapy-refractory acute GvHD in adult and pediatric patients. Long-term data in pediatric patients are scarce. In this retrospective analysis, a total of 25 patients with a median age of 10.6 years (range 0.6-22.1 years) who received bone marrow-derived MSCs after alloHSCT for the treatment of steroid-refractory III and IV GvHD were analyzed. The median observation period of the surviving patients was 9.3 years (1.3-12.7 years) after HSCT. Among the 25 patients, 10 (40.0%) died [relapse (n = 3), multiorgan failure (n = 6), cardiorespiratory failure (n = 1)] at median 0.5 years (0.2-2.3 years) after HSCT. Partial response and complete remission (PR, CR) of the GvHD were achieved in 76.0% and 24.0% of the patients, respectively. Transplant-related mortality was 0% in the patients who achieved CR after MSC treatment and 26.3% for those with PR. A median improvement by one intestinal or liver GvHD stage (range 1-4) could be achieved after MSC application. No potentially MSC-related long-term adverse effects, for example, secondary malignancy, were identified. In conclusion, the intravenous application of allogeneic MSCs was safe and proved effective for the treatment of steroid-refractory GvHD. However, larger, prospective, and randomized trials are needed to evaluate these findings.
Collapse
Affiliation(s)
- Michaela Döring
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | | | - Katrin Lenglinger
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Claudia Treuner
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Friederike Gieseke
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Annika Erbacher
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Markus Mezger
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Martin Vaegler
- Experimental and Clinical Research Center, GMP-Facility for Cellular Therapies, Charité Universitätsmedizin Berlin, Campus Berlin Buch, Berlin, Germany
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology and Oncology, University of Würzburg, Würzburg, Germany
| | - Johann Greil
- Department of Pediatric Hematology and Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Christoph Faul
- University Hospital and Comprehensive Cancer Center Tübingen, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Michael Schumm
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Peter Lang
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department I-General Pediatrics, Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Ingo Müller
- Division for Pediatric Stem Cell Transplantation and Immunology, Clinic for Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Usunier B, Brossard C, L’Homme B, Linard C, Benderitter M, Milliat F, Chapel A. HGF and TSG-6 Released by Mesenchymal Stem Cells Attenuate Colon Radiation-Induced Fibrosis. Int J Mol Sci 2021; 22:ijms22041790. [PMID: 33670243 PMCID: PMC7916908 DOI: 10.3390/ijms22041790] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/27/2021] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Fibrosis is a leading cause of death in occidental states. The increasing number of patients with fibrosis requires innovative approaches. Despite the proven beneficial effects of mesenchymal stem cell (MSC) therapy on fibrosis, there is little evidence of their anti-fibrotic effects in colorectal fibrosis. The ability of MSCs to reduce radiation-induced colorectal fibrosis has been studied in vivo in Sprague–Dawley rats. After local radiation exposure, rats were injected with MSCs before an initiation of fibrosis. MSCs mediated a downregulation of fibrogenesis by a control of extra cellular matrix (ECM) turnover. For a better understanding of the mechanisms, we used an in vitro model of irradiated cocultured colorectal fibrosis in the presence of human MSCs. Pro-fibrotic cells in the colon are mainly intestinal fibroblasts and smooth muscle cells. Intestinal fibroblasts and smooth muscle cells were irradiated and cocultured in the presence of unirradiated MSCs. MSCs mediated a decrease in profibrotic gene expression and proteins secretion. Silencing hepatocyte growth factor (HGF) and tumor necrosis factor-stimulated gene 6 (TSG-6) in MSCs confirmed the complementary effects of these two genes. HGF and TSG-6 limited the progression of fibrosis by reducing activation of the smooth muscle cells and myofibroblast. To settle in vivo the contribution of HGF and TSG-6 in MSC-antifibrotic effects, rats were treated with MSCs silenced for HGF or TSG-6. HGF and TSG-6 silencing in transplanted MSCs resulted in a significant increase in ECM deposition in colon. These results emphasize the potential of MSCs to influence the pathophysiology of fibrosis-related diseases, which represent a challenging area for innovative treatments.
Collapse
|
15
|
Müller L, Tunger A, Wobus M, von Bonin M, Towers R, Bornhäuser M, Dazzi F, Wehner R, Schmitz M. Immunomodulatory Properties of Mesenchymal Stromal Cells: An Update. Front Cell Dev Biol 2021; 9:637725. [PMID: 33634139 PMCID: PMC7900158 DOI: 10.3389/fcell.2021.637725] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/19/2021] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are characterized by an extraordinary capacity to modulate the phenotype and functional properties of various immune cells that play an essential role in the pathogenesis of inflammatory disorders. Thus, MSCs efficiently impair the phagocytic and antigen-presenting capacity of monocytes/macrophages and promote the expression of immunosuppressive molecules such as interleukin (IL)-10 and programmed cell death 1 ligand 1 by these cells. They also effectively inhibit the maturation of dendritic cells and their ability to produce proinflammatory cytokines and to stimulate potent T-cell responses. Furthermore, MSCs inhibit the generation and proinflammatory properties of CD4+ T helper (Th)1 and Th17 cells, while they promote the proliferation of regulatory T cells and their inhibitory capabilities. MSCs also impair the expansion, cytokine secretion, and cytotoxic activity of proinflammatory CD8+ T cells. Moreover, MSCs inhibit the differentiation, proliferation, and antibody secretion of B cells, and foster the generation of IL-10-producing regulatory B cells. Various cell membrane-associated and soluble molecules essentially contribute to these MSC-mediated effects on important cellular components of innate and adaptive immunity. Due to their immunosuppressive properties, MSCs have emerged as promising tools for the treatment of inflammatory disorders such as acute graft-versus-host disease, graft rejection in patients undergoing organ/cell transplantation, and autoimmune diseases.
Collapse
Affiliation(s)
- Luise Müller
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Antje Tunger
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Manja Wobus
- Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Malte von Bonin
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Russell Towers
- Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Martin Bornhäuser
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Francesco Dazzi
- School of Cancer and Pharmacological Sciences and KHP Cancer Research UK Centre, King's College London, London, United Kingdom
| | - Rebekka Wehner
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| |
Collapse
|
16
|
Strobl J, Pandey RV, Krausgruber T, Kleissl L, Reininger B, Herac M, Bayer N, Krall C, Wohlfarth P, Mitterbauer M, Kalhs P, Rabitsch W, Bock C, Hopfinger G, Stary G. Anti-Apoptotic Molecule BCL2 Is a Therapeutic Target in Steroid-Refractory Graft-Versus-Host Disease. J Invest Dermatol 2020; 140:2188-2198. [PMID: 32247860 DOI: 10.1016/j.jid.2020.02.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/11/2020] [Accepted: 02/23/2020] [Indexed: 12/12/2022]
Abstract
Graft-versus-host disease (GVHD) is the leading cause of mortality after hematopoietic stem cell transplantation and primarily affects barrier organs such as the skin. One-third of cases are refractory to steroid treatment resulting in poor outcomes and the need for novel therapies. Longitudinal analysis of T-cell transcriptomes in patients before the appearance of GVHD symptoms revealed the upregulation of anti-apoptotic regulator B-cell lymphoma 2 (BCL2) at GVHD initiation. To determine the potential of BCL2 inhibition in active GVHD, we analyzed tissues of 88 patients with acute or chronic GVHD. BCL2 RNA was elevated in multiple organs affected by GVHD and expression correlated with transplant-related mortality and steroid-refractory GVHD. BCL2-expressing lymphocytes were present in skin lesions and peripheral blood of patients with acute and chronic GVHD. Inhibition of BCL2 increased the CD4 to CD8 ratio in allogeneic T cells in vitro and induced apoptosis of T cells from patients with steroid-pretreated chronic GVHD ex vivo. In addition, the higher ratio of regulatory to nonregulatory T cells upon blockage of BCL2 could add to the anti-inflammatory effect of BCL2 blockage. Collectively, our results highlight BCL2 as an important factor for GVHD development and introduce BCL2 inhibition as previously unreported and urgently needed targeted therapy in the treatment of steroid-refractory GVHD.
Collapse
Affiliation(s)
- Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ram Vinay Pandey
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lisa Kleissl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bärbel Reininger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Merima Herac
- Clinical Institute for Pathology, Medical University of Vienna, Vienna, Austria
| | - Nadine Bayer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Krall
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Austria
| | - Philipp Wohlfarth
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Margit Mitterbauer
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Kalhs
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Werner Rabitsch
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg Hopfinger
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.
| |
Collapse
|
17
|
Therapeutic Effects of Mesenchymal Stem Cells Derived From Bone Marrow, Umbilical Cord Blood, and Pluripotent Stem Cells in a Mouse Model of Chemically Induced Inflammatory Bowel Disease. Inflammation 2020; 42:1730-1740. [PMID: 31227956 DOI: 10.1007/s10753-019-01033-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Acute inflammatory bowel disease (AIBD) is a wide clinical entity including severe gastrointestinal pathologies with common histopathological basis. Epidemiologically increasing diseases, such as necrotizing enterocolitis (NEC), gastrointestinal graft versus host disease (GVHD), and the primary acute phase of chronic inflammatory bowel disease (CIBD), exhibit a high necessity for new therapeutic strategies. Mesenchymal stem cell (MSC) cellular therapy represents a promising option for the treatment of these diseases. In our study, we comparatively assess the efficacy of human MSCs derived from bone marrow (BM), umbilical cord blood (UCB), human embryonic stem cells (ESCs), or human-induced pluripotent stem cells (iPSCs) in a mouse model of chemically induced acute enterocolitis. The laboratory animals were provided ad libitum potable dextrane sulfate sodium solution (DSS) in order to reproduce an AIBD model and then individually exposed intraperitoneally to MSCs derived from BM (BM-MSCs), UCB (UCB-MSCs), ESCs (ESC-MSCs), or iPSCs (iPSC-MSCs). The parameters used to evaluate the cellular treatment efficacy were the animal survival prolongation and the histopathological-macroscopic picture of bowel sections. Although all categories of mesenchymal stem cells led to statistically significant survival prolongation compared to the control group, significant clinical and histopathological improvement was observed only in mice receiving BM-MSCs and UCB-MSCs. Our results demonstrated that the in vivo anti-inflammatory effect of ESC-MSCs and iPSC-MSCs was inferior to that of UCB-MSCs and BM-MSCs. Further investigation will clarify the potential of ESCs and iPSC-derived MSCs in AIBD treatment.
Collapse
|
18
|
Krüger T, Middeke JM, Stölzel F, Mütherig A, List C, Brandt K, Heidrich K, Teipel R, Ordemann R, Schuler U, Oelschlägel U, Wermke M, Kräter M, Herbig M, Wehner R, Schmitz M, Bornhäuser M, von Bonin M. Reliable isolation of human mesenchymal stromal cells from bone marrow biopsy specimens in patients after allogeneic hematopoietic cell transplantation. Cytotherapy 2019; 22:21-26. [PMID: 31883948 DOI: 10.1016/j.jcyt.2019.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
Isolation of mesenchymal stromal cells (MSCs) from pretreated, hematologic patients is challenging. Especially after allogeneic hematopoietic cell transplantation (HCT), standard protocols using bone marrow aspirates fail to reliably recover sufficient cell numbers. Because MSCs are considered to contribute to processes that mainly affect the outcome after transplantation, such as an efficient lymphohematopoietic recovery, extent of graft-versus-host disease as well as the occurrence of leukemic relapse, it is of great clinical relevance to investigate MSC function in this context. Previous studies showed that MSCs can be isolated by collagenase digestion of large bone fragments of hematologically healthy patients undergoing hip replacement or knee surgeries. We have now further developed this procedure for the isolation of MSCs from hematologic patients after allogeneic HCT by using trephine biopsy specimens obtained during routine examinations. Comparison of aspirates and trephine biopsy specimens from patients after allogeneic HCT revealed a significantly higher frequency of clonogenic MSCs (colony-forming unit-fibroblast [CFU-F]) in trephine biopsy specimens (mean, 289.8 ± standard deviation 322.5 CFU-F colonies/1 × 106 total nucleated cells versus 4.2 ± 9.9; P < 0.0001). Subsequent expansion of functional MSCs isolated from trephine biopsy specimen was more robust and led to a significantly higher yield compared with control samples expanded from aspirates (median, 1.6 × 106; range, 0-2.3 × 107 P0 MSCs versus 5.4 × 104; range, 0-8.9 × 106; P < 0.0001). Using trephine biopsy specimens as MSC source facilitates the investigation of various clinical questions.
Collapse
Affiliation(s)
- Thomas Krüger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Anke Mütherig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Catrin List
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Kalina Brandt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Katharina Heidrich
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Raphael Teipel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Rainer Ordemann
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Ulrich Schuler
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Uta Oelschlägel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Wermke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; University Cancer Centrum (UCC), Early Clinical Trial Unit (ECTU), University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Maik Herbig
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Biotechnology Center, Center for Molecular and Cellular Bioengineering TU Dresden Tatzberg 47-49, Dresden, Germany
| | - Rebekka Wehner
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany; Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany; Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Malte von Bonin
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
19
|
Hejretová L, Čedíková M, Dolejšová M, Vlas T, Jindra P, Lysák D, Holubová M. Comparison of the immunomodulatory effect of single MSC batches versus pooled MSC products. Cell Tissue Bank 2019; 21:119-129. [PMID: 31863261 DOI: 10.1007/s10561-019-09805-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Severe corticosteroid-refractory graft-versus-host-disease (GVHD) is a major non-relapse cause of mortality and morbidity after an allogeneic hematopoietic stem cell transplantation (allo-HSCT). One of the most promising treatment options is using advanced therapy medicinal products based on mesenchymal stem cells (MSCs) immunomodulation ability. The protocols of MSC application differ in many parameters including a source of MSC, a dose, a number of doses or way of preparation of the medicinal product. The process is limited by the need for laborious and expensive manufacturing processes fraught with batch-to-batch variability. In our study, we compared the immunomodulatory effects of different MSC batches versus pooled MSC, specifically the influence on lymphocyte proliferation, the metabolic activity, and the expression of activation markers on T cells. Our goal was to determine whether the effect depends on donor-to-donor heterogeneity and if pooling of MSCs could increase their immunomodulatory ability. All tested batches showed an immunomodulatory effect, with no significant differences between the groups. Our study suggests that immunosuppressive potential is comparable in single batches and pooled products, and the use of products got from individual donors is suitable to treat corticosteroid-refractory GVHD.
Collapse
Affiliation(s)
- L Hejretová
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - M Čedíková
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - M Dolejšová
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - T Vlas
- Institute of Immunology and Allergology, University Hospital and Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - P Jindra
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - D Lysák
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - M Holubová
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic. .,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| |
Collapse
|
20
|
Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019; 4:22. [PMID: 31815001 PMCID: PMC6889290 DOI: 10.1038/s41536-019-0083-6] [Citation(s) in RCA: 1191] [Impact Index Per Article: 198.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
The terms MSC and MSCs have become the preferred acronym to describe a cell and a cell population of multipotential stem/progenitor cells commonly referred to as mesenchymal stem cells, multipotential stromal cells, mesenchymal stromal cells, and mesenchymal progenitor cells. The MSCs can differentiate to important lineages under defined conditions in vitro and in limited situations after implantation in vivo. MSCs were isolated and described about 30 years ago and now there are over 55,000 publications on MSCs readily available. Here, we have focused on human MSCs whenever possible. The MSCs have broad anti-inflammatory and immune-modulatory properties. At present, these provide the greatest focus of human MSCs in clinical testing; however, the properties of cultured MSCs in vitro suggest they can have broader applications. The medical utility of MSCs continues to be investigated in over 950 clinical trials. There has been much progress in understanding MSCs over the years, and there is a strong foundation for future scientific research and clinical applications, but also some important questions remain to be answered. Developing further methods to understand and unlock MSC potential through intracellular and intercellular signaling, biomedical engineering, delivery methods and patient selection should all provide substantial advancements in the coming years and greater clinical opportunities. The expansive and growing field of MSC research is teaching us basic human cell biology as well as how to use this type of cell for cellular therapy in a variety of clinical settings, and while much promise is evident, careful new work is still needed.
Collapse
|
21
|
Lu D, Ma T, Zhou X, Jiang Y, Han Y, Li H. B Lymphocytes Are the Target of Mesenchymal Stem Cells Immunoregulatory Effect in a Murine Graft-versus-Host Disease Model. Cell Transplant 2019; 28:1279-1288. [PMID: 31257911 PMCID: PMC6767898 DOI: 10.1177/0963689719860127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
There is growing clinical interest in the utilization of mesenchymal stem cells (MSCs) in the management of acute graft-versus-host disease (aGvHD), yet the effect of major histocompatibility complexes (MHCs) on B lymphocytes in this process has been less well documented. Working in an MHC fully mismatched murine aGvHD model, we found that MSC co-transfer significantly prolonged the survival time of the recipients. More interestingly, analysis on immunophenotypic profiles of posttransplant splenocytes showed that surface expression of CD69 (an early activation marker) and CD86 (a costimulatory molecule) was suppressed predominantly on donor derived B lymphocytes by MSC infusion. Additionally, mRNA level of interleukin-4, a potent B lymphocyte stimulator, was strikingly reduced from MSC-treated mice, while interleukin-10, the regulatory B lymphocytes inductor, was increased; these may underlie the lesser activation of B lymphocytes. In consistence, depletion of B lymphocytes in the transfusion inoculum further prolonged the survival time of aGvHD mice regardless of MSC administration. Therefore, B lymphocytes played an important role in the development of aGvHD, and they are targets in MSC-regulated immune response cascade in vivo. This study may provide a mechanistic clue for the treatment of human clinical aGvHD.
Collapse
Affiliation(s)
- Di Lu
- Department of Plastic and Reconstructive Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China.,Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, China.,Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, China
| | - Tian Ma
- Department of Plastic and Reconstructive Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - XiangBin Zhou
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, China.,Department of Stomatology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - YanMing Jiang
- Department of Ophthalmology, Rocket Force General Hospital, Beijing, China
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hong Li
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, China.,Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, China
| |
Collapse
|
22
|
Abstract
Three decades of research in hematopoietic stem cell transplantation and HIV/AIDS fields have shaped a picture of immune restoration disorders. This manuscript overviews the molecular biology of interferon networks, the molecular pathogenesis of immune reconstitution inflammatory syndrome, and post-hematopoietic stem cell transplantation immune restoration disorders (IRD). It also summarizes the effects of thymic involution on T cell diversity, and the results of the assessment of diagnostic biomarkers of IRD, and tested targeted immunomodulatory treatments.
Collapse
Affiliation(s)
- Hesham Mohei
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Usha Kellampalli
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
23
|
Avivar-Valderas A, Martín-Martín C, Ramírez C, Del Río B, Menta R, Mancheño-Corvo P, Ortiz-Virumbrales M, Herrero-Méndez Á, Panés J, García-Olmo D, Castañer JL, Palacios I, Lombardo E, Dalemans W, DelaRosa O. Dissecting Allo-Sensitization After Local Administration of Human Allogeneic Adipose Mesenchymal Stem Cells in Perianal Fistulas of Crohn's Disease Patients. Front Immunol 2019; 10:1244. [PMID: 31258526 PMCID: PMC6587893 DOI: 10.3389/fimmu.2019.01244] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022] Open
Abstract
Adipose mesenchymal stem cells (ASC) are considered minimally immunogenic. This is due to the low expression of human leukocyte antigens I (HLA-I), lack of HLA-II expression and low expression of co-stimulatory molecules such as CD40 and CD80. The low rate of observed immunological rejection as well as the immunomodulatory qualities, position ASC as a promising cell-based therapy for the treatment of a variety of inflammatory indications. Yet, few studies have addressed relevant aspects of immunogenicity such as ASC donor-to-patient HLA histocompatibility or assessment of immune response triggered by ASC administration, particularly in the cases of presensitization. The present study aims to assess allo-immune responses in a cohort of Crohn's disease patients administered with allogeneic ASC (darvadstrocel formerly Cx601) for the treatment of complex perianal fistulas. We identified donor-specific antibodies (DSA) generation in a proportion of patients and observed that patients showing preexisting immunity were prone to generating DSA after allogeneic therapy. Noteworthy, naïve patients generating DSA at week 12 (W12) showed a significant reduction in DSA titer at week 52 (W52), whereas DSA titer was reduced in pre-sensitized patients only with no specificities against the donor administered. Remarkably, we did not observe any correlation of DSA generation with ASC therapeutic efficacy. In vitro complement-dependent cytotoxicity (CDC) studies have revealed limited cytotoxic levels based upon HLA-I expression and binding capacity even in pro-inflammatory conditions. We sought to identify CDC coping mechanisms contributing to the limited cytotoxic killing observed in ASC in vitro. We found that ASC express membrane-bound complement regulatory proteins (mCRPs) CD55, CD46, and CD59 at basal levels, with CD46 more actively expressed in pro-inflammatory conditions. We demonstrated that CD46 is a main driver of CDC signaling; its depletion significantly enhances sensitivity of ASC to CDC. In summary, despite relatively high clearance, DSA generation may represent a major challenge for allogeneic cell therapy management. Sensitization may be a significant concern when evaluating re-treatment or multi-donor trials. It is still unknown whether DSA generation could potentially be the consequence of donor-to-patient interaction and, therefore, subsequently link to efficacy or biological activity. Lastly, we propose that CDC modulators such as CD46 could be used to ultimately link CDC specificity with allogeneic cell therapy efficacy.
Collapse
Affiliation(s)
| | | | - Cristina Ramírez
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | - Borja Del Río
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | - Ramón Menta
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | | | | | | | - Julián Panés
- Department of Gastroenterology, Hospital Clínic, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Damián García-Olmo
- Department of Surgery, Hospital U. Fundación Jiménez Díaz, Madrid, Spain
| | - José Luís Castañer
- Department of Immunology, University Hospital Ramon y Cajal, Madrid, Spain
| | - Itziar Palacios
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | - Eleuterio Lombardo
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | | | - Olga DelaRosa
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| |
Collapse
|
24
|
Hinden L, Avner M, Stepensky P, Or R, Almogi-Hazan O. Lymphocyte counts may predict a good response to mesenchymal stromal cells therapy in graft versus host disease patients. PLoS One 2019; 14:e0217572. [PMID: 31188842 PMCID: PMC6561566 DOI: 10.1371/journal.pone.0217572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 05/14/2019] [Indexed: 01/08/2023] Open
Abstract
Steroid-resistant GvHD is one of the most significant causes of mortality following allogeneic Hematopoietic Stem Cell Transplantation (HSCT). Treatment with mesenchymal stromal cells (MSC) seems to be a promising solution, however the results from clinical studies are still equivocal. Better selection of candidate patients and improving monitoring of patients following MSC administration can increase treatment effectiveness. In order to determine which characteristics can be used to predict a good response and better monitoring of patients, blood samples were taken prior to therapy, one week and one month after therapy, from 26 allogeneic HSCT patients whom contracted GvHD and were treated with MSCs. Samples were examined for differential blood counts, bilirubin levels and cell surface markers. Serum cytokine levels were also measured. We found that the level of lymphocytes, in particular T and NK cells, may predict a good response to therapy. A better response was observed among patients who expressed low levels of IL-6 and IL-22, Th17 related cytokines, prior to therapy. Patients with high levels of bilirubin prior to therapy showed a poorer response. The results of this study may facilitate early prediction of success or failure of the treatment, and subsequently, will improve selection of patients for MSC therapy.
Collapse
Affiliation(s)
- Liad Hinden
- Department of Bone Marrow Transplantation, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Mordechai Avner
- Department of Bone Marrow Transplantation, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Polina Stepensky
- Department of Bone Marrow Transplantation, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Reuven Or
- Department of Bone Marrow Transplantation, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Osnat Almogi-Hazan
- Department of Bone Marrow Transplantation, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
25
|
ILCs in hematologic malignancies: Tumor cell killers and tissue healers. Semin Immunol 2019; 41:101279. [PMID: 31200953 DOI: 10.1016/j.smim.2019.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 11/21/2022]
Abstract
Innate lymphoid cells (ILCs) have attracted considerable attention in the past years. As modulators of epithelial barrier immunology and homeostasis they play important roles in (auto)immunity and inflammation. Here we review the role of ILCs in hematologic malignancies, where ILCs act as efficient killer cells and as tissue healers, in the context of chemotherapy, radiotherapy and after allogeneic hematopoietic stem cell transplantation (HSCT).
Collapse
|
26
|
Imataki O, Uchida S, Uemura M, Kadowaki N. Graft failure after reduced-intensity stem cell transplantation for congenital sideroblastic anemia. Pediatr Hematol Oncol 2019; 36:46-51. [PMID: 30912988 DOI: 10.1080/08880018.2019.1578844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Congenital sideroblastic anemia (CSA) is a rare hereditary disease causing disorders of hemoglobin synthesis. Severe, progressive congenital sideroblastic anemia becomes transfusion dependent and results in iron overload. In such cases, patients must undergo stem cell transplantation (SCT) before critical organ dysfunction occurs. However, there has been no consensus on the criteria for SCT for congenital sideroblastic anemia. A 17-year-old Japanese boy was newly diagnosed with congenital sideroblastic anemia manifesting dyspnea on effort. His gene analysis revealed ALAS2 R170L. He gradually become dependent on RBC transfusion. Vitamin B6 (pyridoxal, 30 mg/day) therapy and high-dose alpha-linoleic acid supplementation (150 mg/day) had not been effective. We treated the patient with reduced-intensity SCT from a human leukocyte antigen (HLA) alle 8/8-identical unrelated female donor. The preparation regimen applied consisted of cyclophosphamide, fludarabine, total body irradiation (2 Gy), and anti-thymocyte globulin. We experienced secondary graft failure, nevertheless we used enough immunosuppression. Here we discuss the optimal transplantation regimen for an adult-onset congenital sideroblastic anemia patient with transfusion dependency and mild hemosiderosis. We consider a positive indication for SCT in younger (< 20-year-old) patients with congenital sideroblastic anemia with transfusion dependency. Each case should be individually considered for their suitability for SCT depending on the feasibility and the clinical condition of the patient.
Collapse
Affiliation(s)
- Osamu Imataki
- a Division of Hematology, Department of Internal Medicine, Faculty of Medicine , Kagawa University , Kagawa , Japan
| | - Shumpei Uchida
- a Division of Hematology, Department of Internal Medicine, Faculty of Medicine , Kagawa University , Kagawa , Japan
| | - Makiko Uemura
- a Division of Hematology, Department of Internal Medicine, Faculty of Medicine , Kagawa University , Kagawa , Japan
| | - Norimitsu Kadowaki
- a Division of Hematology, Department of Internal Medicine, Faculty of Medicine , Kagawa University , Kagawa , Japan
| |
Collapse
|
27
|
Assessment of the Immunosuppressive Potential of INF-γ Licensed Adipose Mesenchymal Stem Cells, Their Secretome and Extracellular Vesicles. Cells 2019; 8:cells8010022. [PMID: 30621275 PMCID: PMC6356584 DOI: 10.3390/cells8010022] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/25/2018] [Accepted: 12/29/2018] [Indexed: 12/11/2022] Open
Abstract
There is an active search for the ideal strategy to potentialize the effects of Mesenchymal Stem-Cells (MSCs) over the immune system. Also, part of the scientific community is seeking to elucidate the therapeutic potential of MSCs secretome and its extracellular vesicles (EVs), in order to avoid the complexity of a cellular therapy. Here, we investigate the effects of human adipose MSCs (AMSCs) licensing with INF-γ and TLR3 agonist over AMSCs proliferation, migration, as well as the immunomodulatory function. Furthermore, we evaluated how the licensing of AMSCs affected the immunomodulatory function of AMSC derived-secretome, including their EVs. INF-γ licensed-AMSCs presented an elevated expression of indoleamine 2,3-dioxygenase (IDO), accompanied by increased ICAM-1, as well as a higher immunosuppressive potential, compared to unlicensed AMSCs. Interestingly, the conditioned medium obtained from INF-γ licensed-AMSCs also revealed a slightly superior immunosuppressive potential, compared to other licensing strategies. Therefore, unlicensed and INF-γ licensed-AMSCs groups were used to isolate EVs. Interestingly, EVs isolated from both groups displayed similar capacity to inhibit T-cell proliferation. EVs isolated from both groups shared similar TGF-β and Galectin-1 mRNA content but only EVs derived from INF-γ licensed-AMSCs expressed IDO mRNA. In summary, we demonstrated that INF-γ licensing of AMSCs provides an immunosuppressive advantage both from a cell-cell contact-dependent perspective, as well as in a cell-free context. Interestingly, EVs derived from unlicensed and INF-γ licensed-AMSCs have similar ability to control activated T-cell proliferation. These results contribute towards the development of new strategies to control the immune response based on AMSCs or their derived products.
Collapse
|
28
|
Longoni A, Knežević L, Schepers K, Weinans H, Rosenberg AJWP, Gawlitta D. The impact of immune response on endochondral bone regeneration. NPJ Regen Med 2018; 3:22. [PMID: 30510772 PMCID: PMC6265275 DOI: 10.1038/s41536-018-0060-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/26/2018] [Indexed: 12/29/2022] Open
Abstract
Tissue engineered cartilage substitutes, which induce the process of endochondral ossification, represent a regenerative strategy for bone defect healing. Such constructs typically consist of multipotent mesenchymal stromal cells (MSCs) forming a cartilage template in vitro, which can be implanted to stimulate bone formation in vivo. The use of MSCs of allogeneic origin could potentially improve the clinical utility of the tissue engineered cartilage constructs in three ways. First, ready-to-use construct availability can speed up the treatment process. Second, MSCs derived and expanded from a single donor could be applied to treat several patients and thus the costs of the medical interventions would decrease. Finally, it would allow more control over the quality of the MSC chondrogenic differentiation. However, even though the envisaged clinical use of allogeneic cell sources for bone regeneration is advantageous, their immunogenicity poses a significant obstacle to their clinical application. The aim of this review is to increase the awareness of the role played by immune cells during endochondral ossification, and in particular during regenerative strategies when the immune response is altered by the presence of implanted biomaterials and/or cells. More specifically, we focus on how this balance between immune response and bone regeneration is affected by the implantation of a cartilaginous tissue engineered construct of allogeneic origin.
Collapse
Affiliation(s)
- A Longoni
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,Regenerative Medicine Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - L Knežević
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,3Faculty of Health Sciences, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD UK
| | - K Schepers
- 4Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - H Weinans
- 5Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands.,6Department of Rheumatology, University Medical Center Utrecht, Utrecht University, 3584CX Utrecht, The Netherlands.,7Department of Biomechanical Engineering, Delft University of Technology, 2628CD Delft, The Netherlands
| | - A J W P Rosenberg
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands
| | - D Gawlitta
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,Regenerative Medicine Center Utrecht, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
29
|
Featherall J, Robey PG, Rowe DW. Continuing Challenges in Advancing Preclinical Science in Skeletal Cell-Based Therapies and Tissue Regeneration. J Bone Miner Res 2018; 33:1721-1728. [PMID: 30133922 PMCID: PMC6691896 DOI: 10.1002/jbmr.3578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/17/2018] [Accepted: 08/17/2018] [Indexed: 12/28/2022]
Abstract
Cell-based therapies hold much promise for musculoskeletal medicine; however, this rapidly growing field faces a number of challenges. Few of these therapies have proven clinical benefit, and an insufficient regulatory environment has allowed for widespread clinical implementation without sufficient evidence of efficacy. The technical and biological complexity of cell-based therapies has contributed to difficulties with reproducibility and mechanistic clarity. In order to aid in addressing these challenges, we aim to clarify the key issues in the preclinical cell therapy field, and to provide a conceptual framework for advancing the state of the science. Broadly, these suggestions relate to: (i) delineating cell-therapy types and moving away from "catch-all" terms such as "stem cell" therapies; (ii) clarifying descriptions of cells and their processing; and (iii) increasing the standard of in vivo evaluation of cell-based therapy experiments to determining cell fates. Further, we provide an overview of methods for experimental evaluation, data sharing, and professional society participation that would be instrumental in advancing this field. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Joseph Featherall
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA.,Medical Research Scholars Program, Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda MD, USA.,Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda MD, USA
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda MD, USA
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Development, UConn School of Dental Medicine, Farmington, CT, USA
| |
Collapse
|
30
|
Slukvin II, Kumar A. The mesenchymoangioblast, mesodermal precursor for mesenchymal and endothelial cells. Cell Mol Life Sci 2018; 75:3507-3520. [PMID: 29992471 PMCID: PMC6328351 DOI: 10.1007/s00018-018-2871-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/29/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022]
Abstract
Mesenchymoangioblast (MB) is the earliest precursor for endothelial and mesenchymal cells originating from APLNR+PDGFRα+KDR+ mesoderm in human pluripotent stem cell cultures. MBs are identified based on their capacity to form FGF2-dependent compact spheroid colonies in a serum-free semisolid medium. MBs colonies are composed of PDGFRβ+CD271+EMCN+DLK1+CD73- primitive mesenchymal cells which are generated through endothelial/angioblastic intermediates (cores) formed during first 3-4 days of clonogenic cultures. MB-derived primitive mesenchymal cells have potential to differentiate into mesenchymal stromal/stem cells (MSCs), pericytes, and smooth muscle cells. In this review, we summarize the specification and developmental potential of MBs, emphasize features that distinguish MBs from other mesenchymal progenitors described in the literature and discuss the value of these findings for identifying molecular pathways leading to MSC and vasculogenic cell specification, and developing cellular therapies using MB-derived progeny.
Collapse
Affiliation(s)
- Igor I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin, 1220 Capitol Ct., Madison, WI, 53715, USA.
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53707, USA.
- Department of Pathology and Laboratory Medicine, University of Wisconsin, 1685 Highland Ave, Madison, WI, 53705, USA.
| | - Akhilesh Kumar
- Wisconsin National Primate Research Center, University of Wisconsin, 1220 Capitol Ct., Madison, WI, 53715, USA
| |
Collapse
|
31
|
van Hoeven V, Munneke JM, Cornelissen AS, Omar SZ, Spruit MJ, Kleijer M, Bernink JH, Blom B, Voermans C, Hazenberg MD. Mesenchymal Stromal Cells Stimulate the Proliferation and IL-22 Production of Group 3 Innate Lymphoid Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:1165-1173. [PMID: 29980610 DOI: 10.4049/jimmunol.1700901] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 06/07/2018] [Indexed: 12/13/2022]
Abstract
Infusion of mesenchymal stromal cells (MSCs) is a promising and increasingly applied therapy for patients who suffer from a variety of inflammatory diseases, including graft-versus-host disease (GvHD), a common and life-threatening complication after allogeneic hematopoietic stem cell transplantation. The therapeutic effect of MSCs is mainly ascribed to their ability to suppress T cells and to support tissue repair. However, clinical response rates in patients with GvHD are limited to 50%, and the determinants for MSC responsiveness are unknown. We recently reported that high frequencies of activated group 3 innate lymphoid cells (ILC3s) before and after allogeneic hematopoietic stem cell transplantation were associated with a lower risk of GvHD. This may be related to IL-22 production by ILC3s, a cytokine important for intestinal epithelial cell homeostasis. In this study, we investigated whether ILC3s may contribute to the therapeutic effect of MSCs by studying the interaction between MSCs and ILC3s in vitro. ILC3s isolated from human tonsils were cocultured with human bone marrow-derived MSCs for 5 d in the presence of IL-2. Coculture with MSCs enhanced the proliferation and IL-22 production of ILC3s. Reciprocally, ILC3s promoted ICAM-1 and VCAM-1 expression on MSCs. For both directions, the activation was mainly mediated by cell-cell contact and by MSC-derived IL-7 and likely by aryl hydrocarbon receptor ligands. Thus, in addition to inhibiting the proliferation of alloreactive T cells, MSCs also promote the expansion and IL-22 production of ILC3s, which may contribute to healthy homeostasis and wound repair in the treatment of various inflammatory conditions in the intestine, including GvHD.
Collapse
Affiliation(s)
- Vera van Hoeven
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - J Marius Munneke
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Anne S Cornelissen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, 1066 CX Amsterdam, the Netherlands
| | - Said Z Omar
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Melchior J Spruit
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Marion Kleijer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, 1066 CX Amsterdam, the Netherlands
| | - Jochem H Bernink
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Bianca Blom
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, 1066 CX Amsterdam, the Netherlands
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands;
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| |
Collapse
|
32
|
Thielen FW, Blommestein HM, Oosten LEM, Calkoen FG, Lankester AC, Zwaginga JJ, Le Blanc K, Redondo A, Sánchez-Guijo F, Algeri M, Locatelli F, Fibbe WE, Uyl-de Groot CA. Second-line treatment for acute graft-versus-host disease with mesenchymal stromal cells: A decision model. Eur J Haematol 2018; 101:676-683. [PMID: 30084111 DOI: 10.1111/ejh.13158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVE No standard second-line treatment exists for acute graft-versus-host disease steroid-refractory (SR-aGvHD), and long-term outcomes remain poor. Mesenchymal stromal cells (MSCs) have been evaluated as treatment, but no disease model (DM) exists that integrates and extrapolates currently available evidence. The aim of this study was to develop such a DM to describe the natural history of SR-aGvHD and to predict long-term outcomes. METHOD The DM was developed in collaboration with experts in haematology-oncology. Subsequently, a model simulation was run. Input parameters for transition and survival estimates were informed by published data of clinical trials on MSC treatment for SR-aGvHD. Parametric distributions were used to estimate long-term survival rates after MSCs. RESULTS The newly developed DM is a cohort model that consists of eight health states. For the model simulation, we obtained data on 327 patients from 14 published phase II trials. Due to limited evidence, DM structure was simplified and several assumptions had to be made. Median overall survival was 3.2 years for complete response and 0.5 years for no complete response. CONCLUSION The DM provides a comprehensive overview on the second-line treatment pathway for aGvHD and enables long-term predictions that can be used to perform a cost-effectiveness analysis comparing any treatment for SR-aGvHD.
Collapse
Affiliation(s)
- Frederick W Thielen
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
| | - Hedwig M Blommestein
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
- Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Liesbeth E M Oosten
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Friso G Calkoen
- Department of Paediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjan C Lankester
- Department of Paediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap J Zwaginga
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
- Center for Clinical Transfusion Research, Sanquin Research, Leiden, The Netherlands
| | - Katarina Le Blanc
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alba Redondo
- IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain
| | | | | | | | - Wim E Fibbe
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Carin A Uyl-de Groot
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Česen Mazič M, Girandon L, Kneževič M, Avčin SL, Jazbec J. Treatment of Severe Steroid-Refractory Acute-Graft-vs.-Host Disease With Mesenchymal Stem Cells-Single Center Experience. Front Bioeng Biotechnol 2018; 6:93. [PMID: 30087891 PMCID: PMC6066564 DOI: 10.3389/fbioe.2018.00093] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022] Open
Abstract
The most effective treatment of steroid refractory acute graft vs. host disease (aGvHD) is not yet established and mesenchymal stem cells (MSC) appear to be a promising therapy for the condition. We report single center case series of three patients, who underwent allogeneic hematopoietic cell transplantation and later developed steroid refractory graft-vs.-host disease, treated with MSC infusions. Two patients achieved complete remission and one patient partial remission of skin and/or gastrointestinal aGvHD. We demonstrated application of MSC for treatment of severe steroid refractory aGvHD is feasible in clinical practice. Detailed description of patient's features and MSC production protocol is crucial for future comparison on efficacy and safety of cell-based therapies. However, for any substantial conclusions regarding efficacy of MSC higher patient numbers will be required.
Collapse
Affiliation(s)
- Maja Česen Mazič
- Department for Pediatric Hematology and Oncology, University Children Hospital Ljubljana, Ljubljana, Slovenia
| | | | | | - Simona L Avčin
- Department for Pediatric Hematology and Oncology, University Children Hospital Ljubljana, Ljubljana, Slovenia
| | - Janez Jazbec
- Department for Pediatric Hematology and Oncology, University Children Hospital Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
34
|
Teofili L, Silini AR, Bianchi M, Valentini CG, Parolini O. Incorporating placental tissue in cord blood banking for stem cell transplantation. Expert Rev Hematol 2018; 11:649-661. [PMID: 29856650 DOI: 10.1080/17474086.2018.1483717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Human term placenta is comprised of various tissues from which different cells can be obtained, including hematopoietic stem cells and mesenchymal stem/stromal cells (MSCs). Areas covered: This review will discuss the possibility to incorporate placental tissue cells in cord blood banking. It will discuss general features of human placenta, with a brief review of the immune cells at the fetal-maternal interface and the different cell populations isolated from placenta, with a particular focus on MSCs. It will address the question as to why placenta-derived MSCs should be banked with their hematopoietic counterparts. It will discuss clinical trials which are studying safety and efficacy of placenta tissue-derived MSCs in selected diseases, and preclinical studies which have proven their therapeutic properties in other diseases. It will discuss banking of umbilical cord blood and raise several issues for improvement, and the applications of cord blood cells in non-malignant disorders. Expert commentary: Umbilical cord blood banking saves lives worldwide. The concomitant banking of non-hematopoietic cells from placenta, which could be applied therapeutically in the future, alone or in combination to their hematopoietic counterparts, could exploit current banking processes while laying the foundation for clinical trials exploring placenta-derived cell therapies in regenerative medicine.
Collapse
Affiliation(s)
- Luciana Teofili
- a Policlinico Universitario A. Gemelli IRCCS , Banca del Sangue di Cordone Ombelicale UNICATT, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Antonietta R Silini
- b Centro di Ricerca "E. Menni" Fondazione Poliambulanza - Istituto Ospedaliero , Brescia , Italy
| | - Maria Bianchi
- c Policlinico Universitario A. Gemelli IRCCS, Banca del Sangue di Cordone Ombelicale UNICATT , Rome , Italy
| | | | - Ornella Parolini
- b Centro di Ricerca "E. Menni" Fondazione Poliambulanza - Istituto Ospedaliero , Brescia , Italy.,d Istituto di Anatomia Umana e Biologia Cellulare Facoltà di Medicina e chirurgia "A. Gemelli" , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
35
|
Langrzyk A, Nowak WN, Stępniewski J, Jaźwa A, Florczyk-Soluch U, Józkowicz A, Dulak J. Critical View on Mesenchymal Stromal Cells in Regenerative Medicine. Antioxid Redox Signal 2018; 29:169-190. [PMID: 28874054 DOI: 10.1089/ars.2017.7159] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The belief in the potency of stem cells has resulted in the medical applications of numerous cell types for organ repair, often with the low adherence to methodological stringency. Such uncritical enthusiasm is mainly presented in the approaches employing so-called mesenchymal stem cells (MSC), for the treatment of numerous, unrelated conditions. However, it should be stressed that such broad clinical applications of MSC are mostly based on the belief that MSC can efficiently differentiate into multiple cell types, not only osteoblasts, chondrocytes and adipose cells. Recent Advances: Studies employing lineage tracing established more promising markers to characterize MSC identity and localization in vivo and confirmed the differences between MSC isolated from various organs. Furthermore, preclinical and clinical experiments proved that transdifferentiation of MSC is unlikely to contribute to repair of numerous tissues, including the heart. Therefore, the salvage hypotheses, like MSC fusion with cells in target organs or the paracrine mechanisms, were proposed to justify the widespread application of MSC and to explain transient, if any, effects. CRITICAL ISSUES The lack of standardization concerning the cells markers, their origin and particularly the absence of stringent functional characterization of MSC, leads to propagation of the worrying hype despite the lack of convincing therapeutic efficiency of MSC. FUTURE DIRECTIONS The adherence to rigorous methodological rules is necessary to prevent the application of procedures which can be dangerous for patients and scientific research on the medical application of stem cells. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Witold N Nowak
- 2 Cardiovascular Division, King's College London , London, United Kingdom .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Jacek Stępniewski
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Agnieszka Jaźwa
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Urszula Florczyk-Soluch
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Alicja Józkowicz
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Józef Dulak
- 1 Kardio-Med Silesia , Zabrze, Poland .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| |
Collapse
|
36
|
Graft-versus-host diseases characterized by effusion: A case of steroid-refractory graft-versus-host disease. TRANSPLANTATION REPORTS 2018. [DOI: 10.1016/j.tpr.2018.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
37
|
Chen F, Bi D, Cao G, Cheng C, Ma S, Liu Y, Cheng K. Bone morphogenetic protein 7-transduced human dermal-derived fibroblast cells differentiate into osteoblasts and form bone in vivo. Connect Tissue Res 2018; 59:223-232. [PMID: 28696808 DOI: 10.1080/03008207.2017.1353085] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Human dermal-derived fibroblast cells (hDDFCs) are multipotent. Bone morphogenetic proteins (BMPs) are a group of cytokines that promote different developmental processes, including the formation of bone. BMPs can promote hDDFC osteogenesis, but the role of BMP7 in hDDFC osteogenesis in vitro and bone formation in vivo has not been investigated in depth. MATERIALS AND METHODS hDDFCs were stably transfected with a human BMP7 recombinant adenovirus and osteogenic differentiation was examined by alkaline phosphatase staining and calcium accumulation. In addition, we measured the expression of osteoblast-related genes. To examine osteogenesis in vivo, we injected C57BL/6 nude mice with adenovirus-transfected hDDFCs in a calcium alginate hydrogel and examined bone formation using soft X-ray, histological, and immunohistochemical analyses. RESULTS Our findings showed that adenovirus-mediated BMP7 expression promoted osteogenic differentiation of hDDFCs and enhanced expression of osteoblast-related genes in vitro. Cells infected with BMP7 adenoviruses showed enhanced bone formation and osteoblast-related gene expression in vivo after the injection of hDDFC-hydrogel mixture. CONCLUSIONS Taken together, our data indicate that BMP7 significantly promotes hDDFC osteogenesis, and confirm that infecting hDDFCs with BMP7-expressing adenoviruses is a useful tool for bone tissue engineering.
Collapse
Affiliation(s)
- Fuguo Chen
- a Department of Plastic and Reconstructive Surgery , Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Dan Bi
- a Department of Plastic and Reconstructive Surgery , Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Guangqing Cao
- a Department of Plastic and Reconstructive Surgery , Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Chen Cheng
- a Department of Plastic and Reconstructive Surgery , Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Sunxiang Ma
- a Department of Plastic and Reconstructive Surgery , Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Yang Liu
- a Department of Plastic and Reconstructive Surgery , Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Kaixiang Cheng
- a Department of Plastic and Reconstructive Surgery , Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
38
|
Planchon SM, Lingas KT, Reese Koç J, Hooper BM, Maitra B, Fox RM, Imrey PB, Drake KM, Aldred MA, Lazarus HM, Cohen JA. Feasibility of mesenchymal stem cell culture expansion for a phase I clinical trial in multiple sclerosis. Mult Scler J Exp Transl Clin 2018; 4:2055217318765288. [PMID: 29623216 PMCID: PMC5881997 DOI: 10.1177/2055217318765288] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/22/2018] [Indexed: 12/17/2022] Open
Abstract
Background Multiple sclerosis is an inflammatory, neurodegenerative disease of the central nervous system for which therapeutic mesenchymal stem cell transplantation is under study. Published experience of culture-expanding multiple sclerosis patients' mesenchymal stem cells for clinical trials is limited. Objective To determine the feasibility of culture-expanding multiple sclerosis patients' mesenchymal stem cells for clinical use. Methods In a phase I trial, autologous, bone marrow-derived mesenchymal stem cells were isolated from 25 trial participants with multiple sclerosis and eight matched controls, and culture-expanded to a target single dose of 1-2 × 106 cells/kg. Viability, cell product identity and sterility were assessed prior to infusion. Cytogenetic stability was assessed by single nucleotide polymorphism analysis of mesenchymal stem cells from 18 multiple sclerosis patients and five controls. Results One patient failed screening. Mesenchymal stem cell culture expansion was successful for 24 of 25 multiple sclerosis patients and six of eight controls. The target dose was achieved in 16-62 days, requiring two to three cell passages. Growth rate and culture success did not correlate with demographic or multiple sclerosis disease characteristics. Cytogenetic studies identified changes on one chromosome of one control (4.3%) after extended time in culture. Conclusion Culture expansion of mesenchymal stem cells from multiple sclerosis patients as donors is feasible. However, culture time should be minimized for cell products designated for therapeutic administration.
Collapse
Affiliation(s)
| | - Karen T Lingas
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Jane Reese Koç
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Brittney M Hooper
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Basabi Maitra
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Robert M Fox
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Peter B Imrey
- Mellen Center, Neurological Institute, USA.,Quantitative Health Sciences, Lerner Research Institute, USA
| | - Kylie M Drake
- Genomic Medicine Institute, Lerner Research Institute, USA
| | | | - Hillard M Lazarus
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | | |
Collapse
|
39
|
Broglie L, Rademaker A, Galvin J, Ray A, Tse WT, Duerst R, Schneiderman J, Kletzel M, Chaudhury S. Fecal calprotectin and serum albumin as markers of gastrointestinal graft versus host disease. Hematol Oncol Stem Cell Ther 2018; 11:169-174. [PMID: 29397331 DOI: 10.1016/j.hemonc.2017.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/30/2017] [Accepted: 12/26/2017] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Acute graft versus host disease (aGVHD) affects approximately 30-60% of patients after allogeneic hematopoietic stem cell transplantation (HCT) and our ability to predict who develops this complication and their response to treatment is limited. Fecal calprotectin has recently gained popularity as an effective marker of GI inflammation in patients with Inflammatory Bowel Disease (IBD). METHODS Fecal calprotectin and albumin were evaluated as prognostic and predictive markers of aGVHD in 60 adult and pediatric HCT patients. Stool samples were sent for calprotectin quantification prior to starting conditioning, at day 14 post-HCT, at day 28 post-HCT, and at onset of aGVHD ± 2 days. RESULTS Fecal calprotectin did not differentiate patients with GI-GVHD and non-GI GVHD and did not vary based on severity. However, in patients with steroid-refractory GI aGVHD, significantly higher fecal calprotectin levels were noted. At onset of lower-GI symptoms, steroid refractory patients (n = 3) had a mean fecal calprotectin level of 449 ug/g (range 116-1111 ug/g) and a mean albumin of 1.93 g/dL (range 1.6-2.3 g/dL) compared with a mean fecal calprotectin of 24 ug/g (range 16-31 ug/g) and a mean albumin of 3.3 g/dL (range 2.3-3.9 g/dL) in steroid responsive patients (n = 9) (fecal calprotectin p = 0.032, albumin p = 0.027). CONCLUSION Patients with steroid-refractory GI aGVHD had higher fecal calprotectin levels and lower albumin levels than patients with steroid-responsive disease. We recommend further studies to evaluate non-invasive tests with fecal calprotectin in combination with albumin in predicting steroid refractory disease at onset of symptoms to potentially identify patients that may benefit from upfront escalation in GVHD treatment.
Collapse
Affiliation(s)
- Larisa Broglie
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Alfred Rademaker
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John Galvin
- Department of Medicine, Division of Hematology, Oncology and Stem Cell Transplant, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ayita Ray
- Department of Medicine, Division of Hematology, Oncology and Stem Cell Transplant, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - William T Tse
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Reggie Duerst
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jennifer Schneiderman
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Morris Kletzel
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sonali Chaudhury
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
40
|
Correlation of graft immune composition with outcomes after allogeneic stem cell transplantation: Moving towards a perfect transplant. Cell Immunol 2018; 323:1-8. [DOI: 10.1016/j.cellimm.2017.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
|
41
|
Perico N, Casiraghi F, Remuzzi G. Clinical Translation of Mesenchymal Stromal Cell Therapies in Nephrology. J Am Soc Nephrol 2017; 29:362-375. [PMID: 29191959 DOI: 10.1681/asn.2017070781] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells have emerged as potential candidates for cell-based therapies to modulate the immune response in organ transplantation and repair tissues after acute or chronic injury. Preclinical studies have shown convincingly in rodent models that mesenchymal stromal cells can prolong solid organ graft survival and that they can induce immune tolerance, accelerate recovery from AKI, and promote functional improvement in chronic nephropathies. Multiple complex properties of the cells, including immunomodulatory, anti-inflammatory, and proregenerative effects, seem to contribute. The promising preclinical studies have encouraged investigators to explore the safety, tolerability, and efficacy of mesenchymal stromal cell-based therapy in pilot clinical trials, including those for bone marrow and solid organ transplantation, autoimmune diseases, and tissue and organ repair. Here, we review the available data on culture-expanded mesenchymal stromal cells tested in renal transplantation, AKI, and CKD. We also briefly discuss the relevant issues that must be addressed to ensure rigorous assessment of the safety and efficacy of mesenchymal stromal cell therapies to allow the translation of this research into the practice of clinical nephrology.
Collapse
Affiliation(s)
- Norberto Perico
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy;
| | - Federica Casiraghi
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Giuseppe Remuzzi
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy.,Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy; and.,L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
42
|
Cirillo M, Tan P, Sturm M, Cole C. Cellular Immunotherapy for Hematologic Malignancies: Beyond Bone Marrow Transplantation. Biol Blood Marrow Transplant 2017; 24:433-442. [PMID: 29102721 DOI: 10.1016/j.bbmt.2017.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023]
Abstract
Immunotherapy has changed treatment practices for many hematologic malignancies. Even in the current era of targeted therapy, chemotherapy remains the backbone of treatment for many hematologic malignancies, especially in acute leukemias, where relapse remains the major cause of mortality. Application of novel immunotherapies in hematology attempts to harness the killing power of the immune system against leukemia and lymphoma. Cellular immunotherapy is evolving rapidly for high-risk hematologic disorders. Recent advances include chimeric antigen-receptor T cells, mesenchymal stromal/stem cells, dendritic cell tumor vaccines, cytokine-induced killer cells, and virus-specific T cells. The advantages of nontransplantation cellular immunotherapy include suitability for patients for whom transplantation has failed or is contraindicated, and a potentially less-toxic treatment alternative to transplantation for relapsed/refractory patients. This review examines those emerging cellular immunotherapies that are changing treatment paradigms for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Melita Cirillo
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia.
| | - Peter Tan
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Marian Sturm
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Catherine Cole
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
43
|
Lee HS, Song S, Shin DY, Kim GS, Lee JH, Cho CW, Lee KW, Park H, Ahn C, Yang J, Yang HM, Park JB, Kim SJ. Enhanced effect of human mesenchymal stem cells expressing human TNF-αR-Fc and HO-1 gene on porcine islet xenotransplantation in humanized mice. Xenotransplantation 2017; 25. [DOI: 10.1111/xen.12342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/25/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Han-Sin Lee
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
| | - Sanghyun Song
- Department of Surgery; Dankook University College of Medicine; Dankook University Hospital; Cheonam Korea
| | - Du Yeon Shin
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
- Department of Health Sciences & Technology; Samsung Advanced Institute for Health Sciences & Technology; Graduate School; Sungkyunkwan University; Seoul Korea
| | - Geun-Soo Kim
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
| | - Jong-Hyun Lee
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
| | - Chan Woo Cho
- Department of Surgery; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Kyo Won Lee
- Department of Surgery; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Hyojun Park
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
- Department of Surgery; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Curie Ahn
- Transplantation Center; Seoul National University Hospital; Seoul Korea
| | - Jaeseok Yang
- Transplantation Center; Seoul National University Hospital; Seoul Korea
| | - Heung-Mo Yang
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
- Department of Medicine; Sungkyunkwan University School of Medicine; Kyunggi Korea
| | - Jae Berm Park
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
- Department of Surgery; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Sung-Joo Kim
- Transplantation Research Center; Samsung Biomedical Research Institute; Seoul Korea
- Samsung Medical Center; Stem Cell & Regenerative Medicine Institute; Seoul Korea
- Department of Health Sciences & Technology; Samsung Advanced Institute for Health Sciences & Technology; Graduate School; Sungkyunkwan University; Seoul Korea
- Department of Surgery; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Korea
| |
Collapse
|
44
|
Xie J, Liu B, Chen J, Xu Y, Zhan H, Yang F, Li W, Zhou X. Umbilical cord-derived mesenchymal stem cells alleviated inflammation and inhibited apoptosis in interstitial cystitis via AKT/mTOR signaling pathway. Biochem Biophys Res Commun 2017; 495:546-552. [PMID: 29137981 DOI: 10.1016/j.bbrc.2017.11.072] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 12/31/2022]
Abstract
Interstitial cystitis (IC) is a bladder syndrome characterized by pelvic pain and urinary frequency without infection or other identifiable pathology. There are no effective treatments to cure IC. This study investigated the effects of human umbilical cord-derived mesenchymal stem cells (UC-MSCs) injection on IC rat model. Furthermore, we used a coculture system to find the possible molecular mechanism on the human uroepithelial cells (SV-HUC-1), which was the cell model of IC. A rat model of IC was established via systemic injection with cyclophosphamide (CYP) and a cell model of IC was induced by being exposed to tumor necrosis factor (TNF)-α (10 ng/ml). After one week, UC-MSCs injection significantly ameliorated the bladder voiding function in IC rat model. And the Histo- and immunohistochemical analyses showed that UC-MSCs can repair impaired bladder, reduce mast cell infiltration and inhibit apoptosis of urothelium. ELISA results showed that UC-MSCs can decrease IL-1β, IL-6 and TNF-α in bladder. In the coculture system, UC-MSCs can promote proliferation of impaired SV-HUC-1 cells, and inhibit apoptosis. However, while knocked down EGF secreted by UC-MSCs with siRNA, the effects would be weaken. Western blot showed that UC-MSCs increase protein expression levels of p-AKT and p-mTOR in SV-HUC-1 cells, and decrease the levels of cleaved caspase-3. Taken together, we provide evidence that UC-MSCs therapy can successfully alleviate IC in a preclinical animal Model and cell model by alleviating inflammation, promoting proliferation and inhibiting apoptosis. In addition, we demonstrate that the AKT/mTOR signaling pathway was activated.
Collapse
Affiliation(s)
- Juncong Xie
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Bolong Liu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jialiang Chen
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuancheng Xu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hailun Zhan
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Fei Yang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wenbiao Li
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiangfu Zhou
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
45
|
Audehm S, Krackhardt AM. Specific Adoptive Cellular Immunotherapy in Allogeneic Stem Cell Transplantation. Oncol Res Treat 2017; 40:691-696. [PMID: 29069663 DOI: 10.1159/000484051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents a treatment option for a diversity of advanced hematopoietic malignancies providing hope for long-term responses especially due to immunogenic effects associated with the treatment modality. Despite respectable progress in the field, relapses and/or opportunistic infections are major reasons for the high treatment-related mortality. However, a number of novel immunotherapeutic approaches using defined cell populations have been developed to directly target residual malignant cells as well as defined infectious diseases. We here provide an overview of current adoptive cellular immunotherapies in the context of allo-HSCT and close with an outlook on new directions within the field.
Collapse
Affiliation(s)
- Stefan Audehm
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | |
Collapse
|
46
|
Giebel B, Kordelas L, Börger V. Clinical potential of mesenchymal stem/stromal cell-derived extracellular vesicles. Stem Cell Investig 2017; 4:84. [PMID: 29167805 DOI: 10.21037/sci.2017.09.06] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/27/2017] [Indexed: 12/12/2022]
Abstract
Within the last two decades mesenchymal stem/stromal cells (MSCs) emerged after hematopoietic stem cells as the second most investigated and applied somatic stem cell entity so far. MSCs mediate immunosuppressive as well as pro-regenerative activities. Against the initial assumption, MSCs may not primarily exert their therapeutic functions in a cellular but rather in a paracrine manner. Here, extracellular vesicles (EVs), such as exosomes and microvesicles, have been identified as major mediators of these paracrine effects. Meanwhile, MSC-EVs have been applied to an increasing amount of different animal models and were tested in a patient suffering from steroid-refractory acute graft-versus-host disease (acute GvHD) as well as in a patient cohort with chronic kidney disease. So far, the MSC-EV administration appears to be safe in humans and all tested animal models. Improvements were reported in all settings. Thus, MSC-EVs appear as promising novel therapeutic agents which might help to improve disease associated symptoms in millions of patients. Here, we review some of the milestones in the field, briefly discuss challenges and highlight clinical aspects of acute GvHD and its treatment with MSCs and MSC-EVs.
Collapse
Affiliation(s)
- Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Verena Börger
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
47
|
Teti A, Econs MJ. Osteopetroses, emphasizing potential approaches to treatment. Bone 2017; 102:50-59. [PMID: 28167345 DOI: 10.1016/j.bone.2017.02.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/22/2022]
Abstract
Osteopetroses are a heterogeneous group of rare genetic bone diseases sharing the common hallmarks of reduced osteoclast activity, increased bone mass and high bone fragility. Osteoclasts are bone resorbing cells that contribute to bone growth and renewal through the erosion of the mineralized matrix. Alongside the bone forming activity by osteoblasts, osteoclasts allow the skeleton to grow harmonically and maintain a healthy balance between bone resorption and formation. Osteoclast impairment in osteopetroses prevents bone renewal and deteriorates bone quality, causing atraumatic fractures. Osteopetroses vary in severity and are caused by mutations in a variety of genes involved in bone resorption or in osteoclastogenesis. Frequent signs and symptoms include osteosclerosis, deformity, dwarfism and narrowing of the bony canals, including the nerve foramina, leading to hematological and neural failures. The disease is autosomal, with only one extremely rare form associated so far to the X-chromosome, and can have either recessive or dominant inheritance. Recessive ostepetroses are generally lethal in infancy or childhood, with a few milder forms clinically denominated intermediate osteopetroses. Dominant osteopetrosis is so far associated only with mutations in the CLCN7 gene and, although described as a benign form, it can be severely debilitating, although not at the same level as recessive forms, and can rarely result in reduced life expectancy. Severe osteopetroses due to osteoclast autonomous defects can be treated by Hematopoietic Stem Cell Transplant (HSCT), but those due to deficiency of the pro-osteoclastogenic cytokine, RANKL, are not suitable for this procedure. Likewise, it is unclear as to whether HSCT, which has high intrinsic risks, results in clinical improvement in autosomal dominant osteopetrosis. Therefore, there is an unmet medical need to identify new therapies and studies are currently in progress to test gene and cell therapies, small interfering RNA approach and novel pharmacologic treatments.
Collapse
Affiliation(s)
- Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, via Vetoio-Coppito 2, 67100 L'Aquila, Italy.
| | - Michael J Econs
- Department of Medicine, Indiana University, 1120 W. Michigan St., Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University, 1120 W. Michigan St., Indianapolis, IN 46202, USA.
| |
Collapse
|
48
|
Mesenchymal Stromal Cell Therapy: After the Gold Rush. Transplantation 2017; 102:7-8. [PMID: 28858990 DOI: 10.1097/tp.0000000000001932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Lee MK, Lin SP, HuangFu WC, Yang DS, Liu IH. Endothelial-derived extracellular matrix ameliorate the stemness deprivation during ex vivo expansion of mouse bone marrow-derived mesenchymal stem cells. PLoS One 2017; 12:e0184111. [PMID: 28854282 PMCID: PMC5576725 DOI: 10.1371/journal.pone.0184111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold great potential in cell therapies by virtue of the regenerative effects and immunomodulatory properties, but the scarce nature of MSCs makes ex vivo expansion indispensable prior to transplantation purposes. However, potential loss of stemness ensuing culture expansion has hindered the advancements in MSCs-based treatments. In principle, stemness could be preserved by reconstructing the stem cell niche. To test whether the endothelial cells (ECs) participate in the constitution of the stem cell niche for mesenchymal stem cells (MSCs), ECs derivatives including extracellular matrix (ECM) and conditioned medium (CM) prepared from aortic endothelial cells (AECs) and Mile Sven 1 endothelial cell line (MS1) were investigated for the potential to maintain MSCs stemness. MSCs expanded on endothelial ECMs, especially on MS1-ECM, possessed a more juvenile morphology and showed delayed proliferation, when compared with untreated MSCs and MSCs on MSC-ECM and in CMs. Once induced, MS1-ECM group showed better tri-lineage differentiations indicating that MS1-ECM could better preserve MSC stemness. MSCs on MS1-ECM showed stronger immune-modulatory potential and had significantly higher H3K27me3 with lower Kdm6b expression. Taken together, MS1-ECM shapes an inhibitory chromatin signature and retains MSCs stemness. Our work provided supportive evidence that MSCs can reside in a perivascular niche, and a feasible novel approach for MSCs expansion.
Collapse
Affiliation(s)
- Ming-Kang Lee
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Shau-Ping Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Chun HuangFu
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Dee-Shiuh Yang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- School of Veterinary Medicine, National Taiwan University, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
50
|
Microgrooved-surface topography enhances cellular division and proliferation of mouse bone marrow-derived mesenchymal stem cells. PLoS One 2017; 12:e0182128. [PMID: 28846679 PMCID: PMC5573154 DOI: 10.1371/journal.pone.0182128] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/12/2017] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells’ (MSCs) fate is largely determined by the various topographical features and a range of extracellular matrix (ECM) components present in their niches. Apart from maintaining structural stability, they regulate cell morphology, division, proliferation, migration and differentiation among others. Traditional MSC cultures, which are mainly based on two-dimensional smooth surfaces of culture dishes and plates, do not provide topographical cues similar to in vivo three-dimensional niches, impacting various cellular processes. Therefore, we culture the mouse bone marrow-derived MSCs on microgrooved bearing surface, partially mimicking in vivo reticulated niche, to study its effect on morphology, pluripotency factor-associated stemness, cell division and rate of proliferation. Following culture, morphological features, and MSC-specific marker gene expression, such as CD29, CD44, Sca-1 along with HSC (Haematopoietic stem cell)-specific markers like CD34, CD45, CD11b were evaluated by microscopy and immunophenotyping, respectively. HSC is another type of bone marrow stem cell population, which concertedly interacts with MSC during various functions, including haematopoiesis. In addition, mesenchymal stem cells were further analyzed for gene expression of pluripotency-associated transcription factors such as Oct3/4, Sox-2, Nanog and Myc, as well as differentiated into adipocytes, osteocytes and chondrocytes. Our results show that microgrooved surface-cultured mesenchymal stem cells (MMSCs) expressed higher levels of expected cell surface and pluripotency-associated markers and proliferated more rapidly (2–3×fold) with higher percentage of cells in S/G2-M-phase, consequently giving rise to higher cell yield compared to standard culture flask-grown cells (MSCs), taken as control. Furthermore, both MSCs and MMSCs showed considerable accumulation of intracellular lipid-droplets, higher alkaline phosphatase activity and secretion of extracellular matrix that are characteristics of adipogenesis, osteogenesis and chondrogenesis, respectively.
Collapse
|